WO2013006797A1 - Vaccin contre le virus syncytial respiratoire humain - Google Patents

Vaccin contre le virus syncytial respiratoire humain Download PDF

Info

Publication number
WO2013006797A1
WO2013006797A1 PCT/US2012/045769 US2012045769W WO2013006797A1 WO 2013006797 A1 WO2013006797 A1 WO 2013006797A1 US 2012045769 W US2012045769 W US 2012045769W WO 2013006797 A1 WO2013006797 A1 WO 2013006797A1
Authority
WO
WIPO (PCT)
Prior art keywords
oil
ammonium chloride
poloxamer
less
ether
Prior art date
Application number
PCT/US2012/045769
Other languages
English (en)
Inventor
Ali I. Fattom
Nicolas LUKACS
James R. Baker
Tarek Hamouda
Vira BITKO
Original Assignee
Nanobio Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanobio Corporation filed Critical Nanobio Corporation
Priority to JP2014519070A priority Critical patent/JP2014520805A/ja
Priority to CA2840982A priority patent/CA2840982C/fr
Priority to EP12735418.1A priority patent/EP2729169A1/fr
Publication of WO2013006797A1 publication Critical patent/WO2013006797A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/722Chitin, chitosan
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/44Oils, fats or waxes according to two or more groups of A61K47/02-A61K47/42; Natural or modified natural oils, fats or waxes, e.g. castor oil, polyethoxylated castor oil, montan wax, lignite, shellac, rosin, beeswax or lanolin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5115Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1027Paramyxoviridae, e.g. respiratory syncytial virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55583Polysaccharides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18521Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present application relates to the field of immunology, in particular, a vaccine composition of the human respiratory syncytial virus (HRSV) strain, L19 (HRSV-L19) that is a hyperproducer of the structural Fusion (F) and Glycoprotein (G) viral proteins, and the use of HRSV-L19 as a vaccine against HRSV infections.
  • HRSV-L19 human respiratory syncytial virus
  • L19 L19
  • F structural Fusion
  • G Glycoprotein
  • the application further relates to the combination of HRSV-L19 with a nanoemulsion, which is a potent immune enhancer, to induce a protective immune response and avoid vaccine-induce disease enhancement.
  • Respiratory Syncytial Virus is a leading cause of serious respiratory disease in young children and the elderly worldwide and there is no vaccine available against this pathogen.
  • Human respiratory syncytial virus (HRSV) infection commonly results in bronchiolitis and is the leading cause for infant hospitalization in the developed countries.
  • HRSV is increasingly being described as a major pathogen in the elderly, transplant patients, and chronic obstructive pulmonary disease (COPD) patients (ref 1 ).
  • U.S. Patent No. 6,015,832 is directed to methods of inactivating Gram-positive bacteria, a bacterial spore, or Gram-negative bacteria.
  • the methods comprise contacting the Gram-positive bacteria, bacterial spore, or Gram-negative bacteria with a bacteria-inactivating (or bacterial-spore inactivating) emulsion.
  • U.S. Patent No. 6,506,803 is directed to methods of killing or neutralizing microbial agents (e.g., bacterial, virus, spores, fungus, on or in humans using an emulsion.
  • 6,559,189 is directed to methods for decontaminating a sample (human, animal, food, medical device, etc.) comprising contacting the sample with a nanoemulsion.
  • the nanoemulsion when contacted with bacteria, virus, fungi, protozoa or spores, kills or disables the pathogens.
  • the antimicrobial when contacted with bacteria, virus, fungi, protozoa or spores, kills or disables the pathogens.
  • nanoemulsion comprises a quaternary ammonium compound, one of
  • composition 1 comprises an emulsion that is antimicrobial against bacteria, virus, fungi, protozoa, and spores.
  • the emulsions comprise an oil and a quaternary ammonium compound.
  • U.S. Patent No. 7,314,624 is directed to methods of inducing an immune response to an immunogen comprising treating a subject via a mucosal surface with a
  • US-2005-0208083-A1 and US-2006-0251684-A1 are directed to nanoemulsions having droplets with preferred sizes.
  • US-2007-0054834-A1 is directed to compositions comprising quaternary ammonium halides and methods of using the same to treat infectious conditions. The quaternary ammonium compound may be provided as part of an emulsion.
  • US-2007-0036831 - A1 is directed to nanoemulsions comprising an anti-inflammatory agent.
  • the present invention provides a novel approach for inducing a protective immune response against HRSV infection by the isolation of a HRSV viral strain which is a hyperproducer of the pivotal immunogenic viral structural proteins, F and G proteins. Having a vaccine candidate that produces higher levels of F protein in its native state within the confines of the normal viral replication cycle is seminal for its usage as an immunogen, as ample amount and proper
  • conformational epitopes will be presented for the generation of neutralizing antibodies and further induction of the protective cellular arm of the immune response.
  • the inventors have succeeded in cultivating HRSV-L19 and demonstrating that the viral strain is a hyperproducer of F and G viral proteins when compared to the commonly used RSV viral strain A2.
  • the more than 2-fold greater levels of the immunogenic F and G protein found within HRSV-L19 is a novel observation which allows for the use of either attenuated or inactivated virus as a vaccine.
  • the invention encompasses a vaccine composition comprising a purified respiratory syncytial virus (RSV) strain L19 (RSV-L19).
  • RSV-L19 virus is a hyperproducer of Fusion (F) and Glycoprotein (G) structural proteins associated with viral particles.
  • the RSV-L19 virus is attenuated human respiratory syncytial virus (HRSV) strain L19.
  • the vaccine composition comprises a human respiratory syncytial virus deposited with the American Type Culture Collection (ATCC) as HRSV-L19.
  • a method for preparing an immunogenic preparation whereby HRSV-L19 is genetically engineered with attenuating mutations and deletions resulting in an attenuating phenotype.
  • the resulting attenuated virus is cultured in an appropriate cell line and harvested.
  • the harvested virus is then purified free from cellular and serum components.
  • the purified virus is then mixed in an acceptable pharmaceutical carrier for use a vaccine composition.
  • vaccine compositions comprising an RSV viral genome (such as RSV strain L19) comprising at least one attenuating mutation.
  • the vaccine compositions comprise an RSV viral genome (such as RSV strain L19) comprising nucleotide modifications denoting attenuating phenotypes.
  • a method for enhancing immunity to human respiratory syncytial virus infections comprising administering to a subject a nanoemulsion formulation comprising HRSV-L19.
  • Another embodiment of the invention is directed to a method for inducing an enhanced immunity against disease caused by human respiratory syncytial virus comprising the step of administering to a subject an effective amount of a purified HRSV-L19 vaccine composition.
  • the subject can produce a protective immune response after at least a single administration of the nanoemulsion vaccine.
  • the immune response can be protective against one or more strains of RSV.
  • the induction of enhanced immunity to HRSV is dependent upon the presence of optimal levels of antigen.
  • a method for preparing an immunogenic preparation whereby the viral strain HRSV-L19 is cultured in an appropriate cell line and harvested. The harvested virus is concentrated and purified free from cellular and serum components.
  • the purified HRSV-L19 is then inactivated and adjuvanted with a nanoemulsion formulation to provide a non-infectious and immunogenic virus.
  • the simple mixing of a nanoemulsion with a vaccine candidate has been shown to produce both mucosal and system immune response.
  • the mixing of the RSV virion particles with a nanoemulsion results in discrete antigen particles in the oil core of the droplet.
  • the antigen is
  • the RSV vaccines comprise an adjuvant.
  • the adjuvant is a nanoemulsion.
  • the nanoemulsion can comprise an aqueous phase, at least one oil, at least one surfactant, and at least one solvent.
  • the present invention provides methods, compositions and kits for inducing an immune response to RSV in a subject.
  • the methods comprise administering to a subject a nanoemulsion RSV vaccine, wherein the nanoemulsion RSV vaccine comprises droplets having an average diameter of less than about 1000 nm.
  • the nanoemulsion RSV vaccine can further comprise an aqueous phase, at least one oil, at least one surfactant, at least one organic solvent, at least one RSV immunogen, and optionally at least one chelating agent.
  • the nanoemulsion RSV vaccine may be administered via any pharmaceutically acceptable method, including but not limited to intranasally.
  • the nanoemulsion RSV vaccine lacks an organic solvent.
  • additional adjuvants may be added to the nanoemulsion RSV vaccine.
  • the RSV vaccines may be formulated as a liquid dispersion, gel, aerosol, pulmonary aerosol, nasal aerosol, ointment, cream, or solid dose.
  • the RSV vaccines may be administered via any pharmaceutically acceptable method, such as parenterally, orally or intranasally.
  • the parenteral administration can be by subcutaneous, intraperitoneal or intramuscular injection.
  • the nanoemulsion and/or nanoemulsion vaccine is not systemically toxic to the subject, produces minimal or no inflammation upon administration, or any combination thereof.
  • the subject undergoes seroconversion after a single administration of the RSV vaccine.
  • the nanoemulsion RSV vaccine composition comprises (a) at least one cationic surfactant; (b) a cationic surfactant which is cetylpyridinium chloride; (c) a cationic surfactant, and wherein the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001 %; (d) a cationic surfactant, and wherein the concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1 .5%, less than about 1 .0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, less than about 0.10%, greater than about 0.001
  • the nanoemulsion RSV vaccine composition comprises (a) at least one cationic surfactant and at least one non- cationic surfactant; (b) at least one cationic surfactant and at least one non- cationic surfactant, wherein the non-cationic surfactant is a nonionic surfactant; (c) at least one cationic surfactant and at least one non-cationic surfactant, wherein the non-cationic surfactant is a polysorbate nonionic surfactant, a poloxamer nonionic surfactant, or a combination thereof; (d) at least one cationic surfactant and at least one nonionic surfactant which is polysorbate 20, polysorbate 80, poloxamer 188, poloxamer 407, or a combination thereof; (e) at least one cationic surfactant and at least one nonionic surfactant which is polysorbate 20, polysorbate 80, poloxamer 188, poloxamer 407, or a combination thereof; (e
  • the RSV vaccines comprise low molecular weight chitosan, medium molecular weight chitosan, high molecular weight chitosan, a glucan, or any combination thereof.
  • the low molecular weight chitosan, median molecular weight chitosan, high molecular weight chitosan, a glucan, or any combination thereof can be present in the nanoemulsion.
  • FIGURES Shows an SDS PAGE of HRSV Infected Cell Lysate (SDS treated) with L19 and A2.
  • Figure 6A shows a 20% nanoemulsion without added antigen.
  • Figure 6B (panel on the right) shows a 20% nanoemulsion combined with 30 g Fluzone®, and illustrates that the HA antigens are located in the oil droplets. The darkly stained antigens are located outside of the
  • nanoemulsion-adjuvanted vaccines in cotton rats (Example 10).
  • the two formulations evaluated include the W 8 o5EC and the
  • Cotton rats received two doses of 30 ⁇ IN of the nanoemulsion-adjuvanted vaccine containing 6.6 g F-ptn. They were challenged with 5x10 5 pfu RSV strain A2 at week 23. Half of the animals were sacrificed at day 4 and half were sacrificed on day 8.
  • Figure 1 1 Shows the results of an immunogenicity study of W 80 P1885EC
  • the Y axis shows the end point titers of specific antibody to F protein and the X axis shows the time period in weeks.
  • the Y-axis shows the serum antibody levels in g/ml and the
  • X-axis shows the time period in weeks.
  • D4 and D8 show the antibody level in the sera after the challenge.
  • FIG. 12 Shows the results of an immunogenicity study of WsosEC
  • nanoemulsion inactivated RSV vaccine in cotton rats The Y axis shows the end point titers of specific antibody to F protein and the X axis shows the time period in weeks.
  • FIG. 13 Shows the immunogenicity of RSV neutralization in cotton rats.
  • Cotton rats were vaccinated with 30 ⁇ of vaccine intranasally, boosted at 4 weeks, and bled at weeks 0, 4, 6, and 8.
  • Neutralization units represent a reciprocal of the highest dilution that resulted in 50% plaque reduction. NEU measurements were performed at 4 weeks (pre boost) and at 6 weeks (2 weeks post boost). Specimens obtained at 6 weeks generated humoral immune responses adequate to allow for NEU analysis. Data is presented as geometric mean with 95% confidence interval (CI) ( Figure 13A). Correlation between EU and NEU is for all animals at 6 weeks using Spearman rho
  • Figure 14 Shows neutralizing antibodies on day 4 and day 8.
  • Figure 14A Shows neutralizing antibodies on day 4 and day 8.
  • Figure 15 Shows the Specific activity of serum antibodies showed that the specific activity (Neutralizing units/ELISA units) of the serum antibodies tends to increase on Day 8 when compared to Day 4 post-challenge.
  • Figure 15A shows the results for W 8 oP-i 88 5EC nanoemulsion combined with RSV strain L19 (NU/EU for the Y axis), at Day 4 and Day 8.
  • Figure 15B shows the results for
  • Figure 16 Shows cross protection at Day 4 for cotton rats that received 3 doses of RSV L19 adjuvanted vaccine, then challenged with RSV strain A2.
  • Figure 16A shows the results for W 80 Pi 88 5EC nanoemulsion combined with RSV strain L19
  • Figure 16B shows the results for W 80 5EC nanoemulsion combined with RSV strain L19.
  • Serum neutralization activity shows equivalent NU against RSV strain L19 or RSV strain A2, demonstrating cross protection between the two RSV strains.
  • Figure 17 Shows viral clearance (RSV strain A2) at Day 4 in lungs of Cotton
  • Vaccinated cotton rats (vaccinated with W 80 Pi 88 5EC nanoemulsion combined with RSV strain L19, or W 80 5EC nanoemulsion combined with RSV strain L19) showed complete clearance of RSV strain A2 challenged virus from the lungs of cotton rats. Na ' fve animals were showing >10 3 pfu RSV strain A2 /gram of lung.
  • Figure 18 Shows IM Cotton rat vaccination and challenge schedule.
  • Figure 19 Shows Serum immune response in cotton rats vaccinated IM with
  • the Y axis shows serum IgG, g/mL, over a 14 week period, at day 4 post-challenge, and at day 8 post-challenge
  • Figure 20 Shows Serum immune response in cotton rats vaccinated IM with
  • Figure 20A shows the end point titers (Y axis) over a 14 week period, at day 4 post-challenge, and at day 8 post-challenge.
  • Figure 20B shows the ELISA units (Y axis) over a 14 week period, at day 4 post-challenge, and at day 8 post- challenge.
  • Figure 21 Shows IM vaccinated cotton rats showed complete clearance of the
  • RSV 4 days following the challenge compared to Na ' fve animals Shows viral clearance (RSV strain A2) at Day 4 in lungs of Cotton Rats.
  • IM vaccinated cotton rats vaccinated with W 8 o5EC nanoemulsion combined with RSV strain L19
  • Na ' fve animals were showing 10 3 pfu RSV strain A2 or greater /gram of lung.
  • Figure 22 Shows the measurement of anti-F antibodies (Y axis) over an 8 week period (X axis) for mice vaccinated either IM or IN with RSV vaccine containing 2x10 5 plaque forming units (PFU) of L19 RSV virus with 1 .7 g of F protein inactivated with 20% W 8 o5EC nanoemulsion adjuvant.
  • PFU plaque forming units
  • PFU plaque forming units
  • Figure 24 Shows measurement of the cytokines IL-4, IL-13, and IL-17 in lung tissue following either IN or IM vaccination of BALB/C mice
  • the present invention provides methods, compositions and kits for the stimulation of an immune response to an RSV immunogen.
  • the present inventors surprisingly discovered that cells infected with RSV L19 virus produce between 3-1 1 fold higher quantities of RSV viral proteins as compared to cells infected with RSV A2 virus (see Example 1 , infra.).
  • the RSV antigen present in the vaccines of the invention is RSV L19 virus, and more preferably human RSV L19 virus, including the purified, attenuated human respiratory syncytial virus (HRSV) strain L19 (HRSV-L19).
  • the HSV viral genome can comprise at least one attenuating mutation, including but not limited to nucleotide
  • RSV L19 strain was found to cause infection and enhanced respiratory disease (ERD) in mice. Moreover, data published showed that it conferred protection without induction of ERD in mice when formulated with nanoemulsion.
  • the RSV Strain L19 isolate was isolated from an RSV-infected infant with respiratory illness in Ann Arbor, Michigan on 3 January 1967 in WI-38 cells and passaged in SPAFAS primary chick kidney cells followed by passage in SPAFAS primary chick lung cells prior to transfer to MRC-5 cells (Herlocher 1999) and subsequently Hep2 cells (Lukacs 2006).
  • Comparison of RSV L19 genome (15,191 -nt; GenBank accession number FJ614813) with the RSV strain A2 (15,222-nt; GenBank accession number M74568) shows that 98% of the genomes are identical. See Example 5. Most coding differences between L19 and A2 are in the F and G genes. Amino acid alignment of the two strains showed that F protein has 14 (97% identical) and G protein has 20 (93% identical) amino acid differences.
  • RSV L19 strain has been demonstrated in animal models to mimic human infection by stimulating mucus production and significant induction of IL-13 using an inoculum of 1 x 10 5 plaque forming units (PFU)/mouse by intra-tracheal administration (Lukacs 2006).
  • PFU plaque forming units
  • RSV L19 Strain NanoBio developed and optimized RSV propagation and purification methods for three viral strains grown in Vera cells and has established multiplicity of Infection (MOI), optimized purification and concentration of the antigen using PEG6000 precipitation and ultracentrifugation.
  • MOI multiplicity of Infection
  • the RSV L19 viral strain is unique in that it produces significantly higher yields of F protein (approximately 10-30 fold more per PFU) than the other strains.
  • F protein content may be a key factor in immunogenicity and the L19 strain currently elicits the most robust immune response.
  • the L19 strain has a shorter propagation time and therefore will be more efficient from a manufacturing perspective.
  • Table 1 1 Example 6.
  • the antibodies generated are highly effective in neutralizing live virus and there is a linear relationship between neutralization and antibody titers. Furthermore, antibodies generated in cotton rats showed cross protection when immunized with the RSV L19 strain and challenged with the RSV A2 strain. Both IM and IN immunization established memory that can be invoked or recalled after an exposure to antigen either as a second boost or exposure to live virus.
  • the methods comprise administering to a subject a nanoemulsion RSV vaccine, wherein the nanoemulsion vaccine comprises droplets having an average diameter of less than about 1000 nm.
  • the nanoemulsion RSV vaccine further comprises (a) an aqueous phase, (b) at least one oil, (c) at least one surfactant, (d) at least one organic solvent, (e) at least one RSV antigen, and (f) optionally comprising at least one chelating agent, or any combination thereof.
  • the nanoemulsion lacks an organic solvent.
  • the subject is selected from adults, elderly subjects, juvenile subjects, infants, high risk subjects, pregnant women, and
  • the nanoemulsion RSV vaccine may be administered intranasally.
  • the nanoemulsion compositions of the invention function as a vaccine adjuvant.
  • Adjuvants serve to: (1 ) bring the antigen—the substance that stimulates the specific protective immune response— into contact with the immune system and influence the type of immunity produced, as well as the quality of the immune response (magnitude or duration); (2) decrease the toxicity of certain antigens; (3) reduce the amount of antigen needed for a protective response; (4) reduce the number of doses required for protection; (5) enhance immunity in poorly responding subsets of the population and/or (7) provide solubility to some vaccines components.
  • the nanoemulsion vaccine adjuvants are particularly useful for adjuvanting RSV vaccines.
  • the RSV vaccines comprise F protein of an RSV strain, such as but not limited to F protein of RSV strain L19. In another embodiment, the RSV vaccines comprise about 0.1 [ ⁇ g up to about 100 [ ⁇ g, and any amount in-between, of RSV F protein, such as F protein of RSV strain L19.
  • the RSV vaccines can comprise about 0.1 [ ⁇ g, about 0.2 [ig, about 0.3 [ig, about 0.4 g, about 0.5 [ig, about 0.6 [ig, about 0.7 g, about 0.8 [ig, about 0.9 [ig, about 1 .0 [ig, about 1 .1 g, about 1 .2 g, about 1 .3 [ig, about 1 .4 about 1 .5 [ig, about 1 .6 [ig, about 1 .7 g, about 1 .8 [ig, about 1 .9 [ig, about 2.0 [ig, about 2.1 pg, about 2.2 pg, about 2.3 [ig, about 2.4 pg, about 2.5 g, about 2.6 g, about 2.7 pg, about 2.8 [ig, about 2.9 g, about 3.0 [ig, about 3.1 g, about 3.2 pg, about 3.3 [ig, about 3.4 pg, about 2.8 [
  • the RSV vaccines of the invention comprise about 1 .0 x 10 5 pfu (plaque forming units (pfu) up to about 1 .0 x 10 8 pfu, and any amount in-between, of an RSV virus, such as RSV strain L19.
  • the RSV virus in inactivated by the presence of the nanoemulsion adjuvant.
  • the RSV vaccines can comprise about 1 .0 x 10 5 , 1 .1 x 10 5 , 1 .2 x 10 5 , 1 .3 x 10 5 ,
  • the RSV vaccines of the invention comprising RSV strain L19 are cross-reactive against at least one other RSV strain (or cross-reactive against one or more RSV strains).
  • cross reactivity can be measured 1 ) using ELISA method to see if the sera from vaccinated animals or individuals will produce antibodies against strains that were not used in the administered vaccine; 2) Immune cells will produce cytokines when stimulated in vitro using stains that were not used in the administered vaccine.
  • Cross protection can be measured in vitro when antibodies in sera of animals vaccinated with one strain will neutralize infectivity of another virus not used in the administered vaccine.
  • the RSV vaccines of the invention comprising RSV strain L19 can be cross reactive against one or more RSV strains selected from the group consisting of RSV strain A2 (wild type) (ATCC VR-1540P), RSV strain rA2cp248/404, RSV Strain 2-20, RSV strain 3-12, RSV strain 58-104, RSV strain Long (ATCC VR-26), RSV strain 9320 (ATCC VR-955), RSV strain B
  • WV/14617/85 (ATCC VR-1400), RSV strain 18537 (ATCC VR-1580), RSV strain A2 cpts-248 (ATCC VR-2450), RSV strain A2 cpts-530/1009 (ATCC VR-2451 ), RSV strain A2 cpts-530 (ATCC VR-2452), RSV strain A2 cpts-248/955 (ATCC VR-2453), RSV strain A2 cpts-248/404 (ATCC VR-2454), RSV strain A2 cpts- 530/1030 (ATCC VR-2455), RSV strain subgroup B cp23 Clone 1A2 (ATCC VR- 2579), RSV strain Subgroup B, Strain B1 , and cp52 Clone 2B5 (ATCC VR-2542).
  • the RSV vaccines of the invention result in a protective immune response following one or two doses of the RSV vaccine.
  • the RSV vaccines of the invention result in generation of robust neutralizing antibodies.
  • the RSV vaccines of the invention result in generation of robust neutralizing antibodies.
  • Administration of one or two doses of an RSV vaccine according to the invention can result in neutralizing antibody titers ranging from 2 to 10 6 or more.
  • Nanoemulsions are oil-in-water emulsions composed of nanometer sized droplets with surfactant(s) at the oil-water interface. Because of their size, the nanoemulsion droplets are pinocytosed by dendritic cells triggering cell maturation and efficient antigen presentation to the immune system. When mixed with different antigens, nanoemulsion adjuvants elicit and up-modulate strong humoral and cellular T H 1 -type responses as well as mucosal immunity (Makidon et al., "Pre-Clinical Evaluation of a Novel Nanoemulsion-Based
  • Vaccine Immunol. 15(2): 348-58 (2008); Warren et al., "Pharmacological and Toxicological Studies on Cetylpyridinium Chloride, A New Germicide," J. Pharmacol. Exp. Ther., 74:401 -8) (1942)).
  • antigens include protective antigen (PA) of anthrax (Bielinska et al., Infect. Immun., 75(8): 4020-9 (2007)), whole vaccinia virus (Bielinska et al., Clin.
  • the nanoemulsion RSV vaccine of the invention can be administered to a subject using any pharmaceutically acceptable method, such as for example, intranasal, buccal, sublingual, oral, rectal, ocular, parenteral (intravenously, intradermally, intramuscularly, subcutaneously, intracisternally, intraperitoneally), pulmonary, intravaginal, locally administered, topically administered, topically administered after scarification, mucosally administered, via an aerosol, or via a buccal or nasal spray formulation.
  • the nanoemulsion RSV vaccine can be formulated into any pharmaceutically acceptable dosage form, such as a liquid dispersion, gel, aerosol, pulmonary aerosol, nasal aerosol, ointment, cream, semi-solid dosage form, or a suspension.
  • the nanoemulsion RSV vaccine may be a controlled release formulation, sustained release formulation, immediate release formulation, or any combination thereof.
  • the nanoemulsion RSV vaccine may be a transdermal delivery system such as a patch or administered by a pressurized or pneumatic device (i.e., a "gene gun").
  • the nanoemulsion RSV vaccine comprises droplets having an average diameter of less than about 1000 nm and: (a) an aqueous phase; (b) about 1 % oil to about 80% oil; (c) about 0.1 % to about 50% organic solvent; (d) about 0.001 % to about 10% of a surfactant or detergent; or (e) any combination thereof.
  • the nanoemulsion vaccine comprises: (a) an aqueous phase; (b) about 1 % oil to about 80% oil; (c) about 0.1 % to about 50% organic solvent; (d) about 0.001 % to about 10% of a surfactant or detergent; and (e) at least one RSV immunogen.
  • the nanoemulsion lacks an organic solvent.
  • the quantities of each component present in the nanoemulsion and/or nanoemulsion vaccine refer to a therapeutic nanoemulsion and/or nanoemulsion RSV vaccine.
  • the nanoemulsion vaccine droplets have an average diameter selected from the group consisting of less than about 1000 nm, less than about 950 nm, less than about 900 nm, less than about 850 nm, less than about 800 nm, less than about 750 nm, less than about 700 nm, less than about 650 nm, less than about 600 nm, less than about 550 nm, less than about 500 nm, less than about 450 nm, less than about 400 nm, less than about 350 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 100 nm, greater than about 50 nm, greater than about 70 nm, greater than about 125 nm, and any combination thereof.
  • the nanoemulsion and/or nanoemulsion vaccine comprises a cationic surfactant which is cetylpyridinium chloride (CPC).
  • CPC may have a concentration in the nanoemulsion RSV vaccine of less than about 5.0% and greater than about 0.001 %, or further, may have a concentration of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1 .5%, less than about 1 .0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, less than about 0.10%, greater than about 0.001 %, greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than
  • the nanoemulsion RSV vaccine comprises a non- ionic surfactant, such as a polysorbate surfactant, which may be polysorbate 80 or polysorbate 20, and may have a concentration of about 0.01 % to about 5.0 %, or about 0.1 % to about 3% of polysorbate 80.
  • the nanoemulsion RSV vaccine may further comprise at least one preservative.
  • the nanoemulsion RSV vaccine comprises a chelating agent.
  • the nanoemulsion RSV vaccine further comprises an immune modulator, such as chitosan or glucan.
  • An immune modulator can be present in the vaccine composition at any pharmaceutically acceptable amount including, but not limited to, from about 0.001 % up to about 10%, and any amount in between, such as about 0.01 %, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1 %, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1 %, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%.
  • the term "adjuvant” refers to an agent that increases the immune response to an antigen (e.g., a pathogen).
  • the term “immune response” refers to a subject's (e.g., a human or another animal) response by the immune system to immunogens (i.e., antigens) which the subject's immune system recognizes as foreign. Immune responses include both cell-mediated immune responses (responses mediated by antigen-specific T cells and non-specific cells of the immune system) and humoral immune responses (responses mediated by antibodies present in the plasma lymph, and tissue fluids).
  • the term “immune response” encompasses both the initial responses to an immunogen (e.g., a pathogen) as well as memory responses that are a result of "acquired immunity.”
  • HRSV refers to viral particles with reduced virulence and pathogenicity and in animal models and human will not result in clinical diseases.
  • chelator or "chelating agent” refer to any materials having more than one atom with a lone pair of electrons that are available to bond to a metal ion.
  • the term "enhanced immunity” refers to an increase in the level of acquired immunity to a given pathogen following administration of a vaccine of the present invention relative to the level of acquired immunity when a vaccine of the present invention has not been administered.
  • hyperproducer refers to a viral strain that is capable of selectively producing at least 2-fold higher levels of viral structural proteins over standard viral strains.
  • hyperproducer refers to the unique demonstration that HRSV-L19 produces levels of F and G proteins that are considerably higher than the comparable A2 HRSV strain.
  • immunogen refers to an antigen that is capable of eliciting an immune response in a subject.
  • immunogens elicit immunity against the immunogen (e.g., a pathogen or a pathogen product) when administered in combination with a nanoemulsion of the present invention.
  • inactivated HRSV refers to virion particles that are incapable of infecting host cells and are noninfectious in pertinent animal models.
  • the term “intranasal(ly)” refers to application of the compositions of the present invention to the surface of the skin and mucosal cells and tissues of the nasal passages, e.g., nasal mucosa, sinus cavity, nasal turbinates, or other tissues and cells which line the nasal passages.
  • nanoemulsion includes small oil-in-water dispersions or droplets, as well as other lipid structures which can form as a result of hydrophobic forces which drive apolar residues (i.e., long hydrocarbon chains) away from water and drive polar head groups toward water, when a water immiscible oily phase is mixed with an aqueous phase.
  • These other lipid structures include, but are not limited to, unilamellar, paucilamellar, and
  • compositions that do not substantially produce adverse allergic or adverse immunological reactions when administered to a host (e.g., an animal or a human).
  • a host e.g., an animal or a human.
  • Such formulations include any
  • pharmaceutically acceptable dosage form examples include, but are not limited to, dips, sprays, seed dressings, stem injections, lyophilized dosage forms, sprays, and mists.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, wetting agents (e.g., sodium lauryl sulfate), isotonic and absorption delaying agents, disintegrants (e.g., potato starch or sodium starch glycolate), and the like.
  • compositions of the present invention to the surface of the skin and mucosal cells and tissues (e.g., buccal, lingual, sublingual, masticatory, respiratory or nasal mucosa, nasal turbinates and other tissues and cells which line hollow organs or body cavities).
  • mucosal cells and tissues e.g., buccal, lingual, sublingual, masticatory, respiratory or nasal mucosa, nasal turbinates and other tissues and cells which line hollow organs or body cavities).
  • viral particles refers to mature virions, partial virions, empty capsids, defective interfering particles, and viral envelopes.
  • the nanoemulsion RSV vaccines of the invention can be stable at about 40°C and about 75% relative humidity for a time period of at least up to about 2 days, at least up to about 2 weeks, at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, or at least up to about 3 years.
  • the nanoemulsion RSV vaccines of the invention can be stable at about 25°C and about 60% relative humidity for a time period of at least up least up to about 2 days, at least up to about 2 weeks, to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, or at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, or at least up to about 5 years.
  • the nanoemulsion RSV vaccines of the invention can be stable at about 4°C for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, at least up to about 5 years, at least up to about 5.5 years, at least up to about 6 years, at least up to about 6.5 years, or at least up to about 7 years.
  • the nanoemulsion RSV vaccines of the invention can be stable at about - 20°C for a time period of at least up to about 1 month, at least up to about 3 months, at least up to about 6 months, at least up to about 12 months, at least up to about 18 months, at least up to about 2 years, at least up to about 2.5 years, at least up to about 3 years, at least up to about 3.5 years, at least up to about 4 years, at least up to about 4.5 years, at least up to about 5 years, at least up to about 5.5 years, at least up to about 6 years, at least up to about 6.5 years, or at least up to about 7 years.
  • the immune response of the subject can be measured by determining the titer and/or presence of antibodies against the RSV immunogen after
  • Seroconversion refers to the development of specific antibodies to an immunogen and may be used to evaluate the presence of a protective immune response.
  • antibody-based detection is often measured using Western blotting or enzyme-linked immunosorbent (ELISA) assays or hemagglutination inhibition assays (HAI). Persons of skill in the art would readily select and use appropriate detection methods.
  • ELISA enzyme-linked immunosorbent
  • HAI hemagglutination inhibition assays
  • Another method for determining the subject's immune response is to determine the cellular immune response, such as through immunogen-specific cell responses, such as cytotoxic T lymphocytes, or immunogen-specific lymphocyte proliferation assay. Additionally, challenge by the pathogen may be used to determine the immune response, either in the subject, or, more likely, in an animal model.
  • immunogen-specific cell responses such as cytotoxic T lymphocytes, or immunogen-specific lymphocyte proliferation assay.
  • challenge by the pathogen may be used to determine the immune response, either in the subject, or, more likely, in an animal model.
  • a person of skill in the art would be well versed in the methods of determining the immune response of a subject and the invention is not limited to any particular method.
  • the nanoemulsion RSV vaccine of the present invention comprises droplets having an average diameter size of less than about 1 ,000 nm, less than about 950 nm, less than about 900 nm, less than about 850 nm, less than about 800 nm, less than about 750 nm, less than about 700 nm, less than about 650 nm, less than about 600 nm, less than about 550 nm, less than about 500 nm, less than about 450 nm, less than about 400 nm, less than about 350 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, or any combination thereof.
  • the droplets have an average diameter size greater than about 125 nm and less than or equal to about 600 nm. In a different embodiment, the droplets have an average diameter size greater than about 50 nm or greater than about 70 nm, and less than or equal to about 125 nm.
  • the aqueous phase can comprise any type of aqueous phase including, but not limited to, water (e.g., H 2 O, distilled water, purified water, water for injection, de-ionized water, tap water) and solutions (e.g., phosphate buffered saline (PBS) solution).
  • the aqueous phase comprises water at a pH of about 4 to 10, preferably about 6 to 8.
  • the water can be deionized (hereinafter "DiH 2 O").
  • the aqueous phase comprises phosphate buffered saline (PBS).
  • the aqueous phase may further be sterile and pyrogen free.
  • Organic solvents in the nanoemulsion RSV vaccines of the invention include, but are not limited to, Ci-C-
  • the organic solvent is an alcohol chosen from a nonpolar solvent, a polar solvent, a protic solvent, or an aprotic solvent.
  • Suitable organic solvents for the nanoemulsion RSV vaccine include, but are not limited to, ethanol, methanol, isopropyl alcohol, glycerol, medium chain triglycerides, diethyl ether, ethyl acetate, acetone, dimethyl sulfoxide (DMSO), acetic acid, n-butanol, butylene glycol, perfumers alcohols, isopropanol, n- propanol, formic acid, propylene glycols, glycerol, sorbitol, industrial methylated spirit, triacetin, hexane, benzene, toluene, diethyl ether, chloroform, 1 ,4-dixoane, tetrahydrofuran, dichloromethane, acetone, acetonitrile, dimethylformamide, dimethyl sulfoxide, formic acid, semi-synthetic derivatives thereof, and any combination thereof. 4. Oil Phase
  • the oil in the nanoemulsion RSV vaccine of the invention can be any cosmetically or pharmaceutically acceptable oil.
  • the oil can be volatile or nonvolatile, and may be chosen from animal oil, vegetable oil, natural oil, synthetic oil, hydrocarbon oils, silicone oils, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable oils include, but are not limited to, mineral oil, squalene oil, flavor oils, silicon oil, essential oils, water insoluble vitamins, Isopropyl stearate, Butyl stearate, Octyl palmitate, Cetyl palmitate, Tridecyl behenate, Diisopropyl adipate, Dioctyl sebacate, Menthyl anthranhilate, Cetyl octanoate, Octyl salicylate, Isopropyl myristate, neopentyl glycol dicarpate cetols, Ceraphyls®, Decyl oleate, diisopropyl adipate, C-
  • the oil may further comprise a silicone component, such as a volatile silicone component, which can be the sole oil in the silicone component or can be combined with other silicone and non-silicone, volatile and non-volatile oils.
  • a silicone component such as a volatile silicone component, which can be the sole oil in the silicone component or can be combined with other silicone and non-silicone, volatile and non-volatile oils.
  • Suitable silicone components include, but are not limited to,
  • methylphenylpolysiloxane simethicone, dimethicone, phenyltrimethicone (or an organomodified version thereof), alkylated derivatives of polymeric silicones, cetyl dimethicone, lauryl trimethicone, hydroxylated derivatives of polymeric silicones, such as dimethiconol, volatile silicone oils, cyclic and linear silicones,
  • cyclomethicone derivatives of cyclomethicone, hexamethylcyclotrisiloxane, octamethylcyclotetrasiloxane, decamethylcyclopentasiloxane, volatile linear dimethylpolysiloxanes, isohexadecane, isoeicosane, isotetracosane, polyisobutene, isooctane, isododecane, semi-synthetic derivatives thereof, and combinations thereof.
  • the volatile oil can be the organic solvent, or the volatile oil can be present in addition to an organic solvent.
  • Suitable volatile oils include, but are not limited to, a terpene, monoterpene, sesquiterpene, carminative, azulene, menthol, camphor, thujone, thymol, nerol, linalool, limonene, geraniol, perillyl alcohol, nerolidol, farnesol, y GmbHe, bisabolol, farnesene, ascaridole, chenopodium oil, citronellal, citral, citronellol, chamazulene, yarrow, guaiazulene, chamomile, semisynthetic derivatives, or combinations thereof.
  • the volatile oil in the silicone component is different than the oil in the oil phase.
  • the surfactant in the nanoemulsion RSV vaccine of the invention can be a pharmaceutically acceptable ionic surfactant, a pharmaceutically acceptable nonionic surfactant, a pharmaceutically acceptable cationic surfactant, a pharmaceutically acceptable anionic surfactant, or a pharmaceutically acceptable zwitterionic surfactant.
  • the surfactant can be a pharmaceutically acceptable ionic polymeric surfactant, a pharmaceutically acceptable nonionic polymeric surfactant, a pharmaceutically acceptable cationic polymeric surfactant, a pharmaceutically acceptable anionic polymeric surfactant, or a pharmaceutically acceptable zwitterionic polymeric surfactant.
  • polymeric surfactants include, but are not limited to, a graft copolymer of a poly(methyl methacrylate) backbone with multiple (at least one) polyethylene oxide (PEO) side chain, polyhydroxystearic acid, an alkoxylated alkyl phenol formaldehyde condensate, a polyalkylene glycol modified polyester with fatty acid hydrophobes, a polyester, semi-synthetic derivatives thereof, or combinations thereof.
  • PEO polyethylene oxide
  • Surface active agents or surfactants are amphipathic molecules that consist of a non-polar hydrophobic portion, usually a straight or branched hydrocarbon or fluorocarbon chain containing 8-18 carbon atoms, attached to a polar or ionic hydrophilic portion.
  • the hydrophilic portion can be nonionic, ionic or zwitterionic.
  • the hydrocarbon chain interacts weakly with the water molecules in an aqueous environment, whereas the polar or ionic head group interacts strongly with water molecules via dipole or ion-dipole interactions.
  • surfactants are classified into anionic, cationic, zwitterionic, nonionic and polymeric surfactants.
  • Suitable surfactants include, but are not limited to, ethoxylated
  • nonylphenol comprising 9 to 10 units of ethyleneglycol, ethoxylated undecanol comprising 8 units of ethyleneglycol, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate, polyoxyethylene (20) sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monostearate, sorbitan monooleate,
  • ethoxylated hydrogenated ricin oils sodium laurylsulfate, a diblock copolymer of ethyleneoxyde and propyleneoxyde, Ethylene Oxide-Propylene Oxide Block Copolymers, and tetra-functional block copolymers based on ethylene oxide and propylene oxide, Glyceryl monoesters, Glyceryl caprate, Glyceryl caprylate, Glyceryl cocate, Glyceryl erucate, Glyceryl hydroxysterate, Glyceryl isostearate, Glyceryl lanolate, Glyceryl laurate, Glyceryl linolate, Glyceryl myristate, Glyceryl oleate, Glyceryl PABA, Glyceryl palmitate, Glyceryl ricinoleate, Glyceryl stearate, Glyceryl thiglycolate, Glyceryl dilaurate, Glyceryl dioleate, Glyceryl
  • Additional suitable surfactants include, but are not limited to, non-ionic lipids, such as glyceryl laurate, glyceryl myristate, glyceryl dilaurate, glyceryl dimyristate, semi-synthetic derivatives thereof, and mixtures thereof.
  • non-ionic lipids such as glyceryl laurate, glyceryl myristate, glyceryl dilaurate, glyceryl dimyristate, semi-synthetic derivatives thereof, and mixtures thereof.
  • the surfactant is a polyoxyethylene fatty ether having a polyoxyethylene head group ranging from about 2 to about 100 groups, or an alkoxylated alcohol having the structure R5 ⁇ (OCH 2 CH 2 ) y -OH, wherein R 5 is a branched or unbranched alkyl group having from about 6 to about 22 carbon atoms and y is between about 4 and about 100, and preferably, between about 10 and about 100.
  • the alkoxylated alcohol is the species wherein R 5 is a lauryl group and y has an average value of 23.
  • the surfactant is an alkoxylated alcohol which is an ethoxylated derivative of lanolin alcohol.
  • the ethoxylated derivative of lanolin alcohol is laneth-10, which is the polyethylene glycol ether of lanolin alcohol with an average ethoxylation value of 10.
  • Nonionic surfactants include, but are not limited to, an ethoxylated surfactant, an alcohol ethoxylated, an alkyl phenol ethoxylated, a fatty acid ethoxylated, a monoalkaolannide ethoxylated, a sorbitan ester ethoxylated, a fatty amino ethoxylated, an ethylene oxide-propylene oxide copolymer,
  • Heptaethylene glycol monodecyl ether Heptaethylene glycol monododecyl ether, Heptaethylene glycol monotetradecyl ether, n-Hexadecyl beta-D-maltoside, Hexaethylene glycol monododecyl ether, Hexaethylene glycol monohexadecyl ether, Hexaethylene glycol monooctadecyl ether, Hexaethylene glycol
  • Octaethylene glycol monodecyl ether Octaethylene glycol monododecyl ether, Octaethylene glycol monohexadecyl ether, Octaethylene glycol monooctadecyl ether, Octaethylene glycol monotetradecyl ether, Octyl-beta-D-glucopyranoside, Pentaethylene glycol monodecyl ether, Pentaethylene glycol monododecyl ether, Pentaethylene glycol monohexadecyl ether, Pentaethylene glycol monohexyl ether, Pentaethylene glycol monooctadecyl ether, Pentaethylene glycol monooctyl ether, Polyethylene glycol diglycidyl ether, Polyethylene glycol ether W-1 , Polyoxyethylene 10 tridecyl ether, Polyoxyethylene 100 stearate,
  • Polyoxyethylene 20 isohexadecyl ether, Polyoxyethylene 20 oleyl ether,
  • nonionic surfactant can be a poloxamer.
  • Poloxamers are polymers made of a block of polyoxyethylene, followed by a block of
  • Poloxamer 101 consists of a block with an average of 2 units of polyoxyethylene, a block with an average of 16 units of polyoxypropylene, followed by a block with an average of 2 units of polyoxyethylene.
  • Poloxamers range from colorless liquids and pastes to white solids. In cosmetics and personal care products, Poloxamers are used in the formulation of skin cleansers, bath products, shampoos, hair conditioners, mouthwashes, eye makeup remover and other skin and hair products.
  • Poloxamers include, but are not limited to, Poloxamer 101 , Poloxamer 105, Poloxamer 108, Poloxamer 122, Poloxamer 123, Poloxamer 124, Poloxamer 181 , Poloxamer 182, Poloxamer 183, Poloxamer 184, Poloxamer 185,
  • Poloxamer 188 Poloxamer 212, Poloxamer 215, Poloxamer 217, Poloxamer 231 , Poloxamer 234, Poloxamer 235, Poloxamer 237, Poloxamer 238,
  • Poloxamer 282 Poloxamer 284, Poloxamer 288, Poloxamer 331 , Poloxamer 333, Poloxamer 334, Poloxamer 335, Poloxamer 338, Poloxamer 401 ,
  • Poloxamer 402 Poloxamer 403, Poloxamer 407, Poloxamer 105 Benzoate, and Poloxamer 182 Dibenzoate.
  • Suitable cationic surfactants include, but are not limited to, a quarternary ammonium compound, an alkyl trimethyl ammonium chloride compound, a dialkyi dimethyl ammonium chloride compound, a cationic halogen-containing
  • Hexadecyltrimethylammonium bromide Hexadecyltrimethylammonium bromide, N,N',N'-Polyoxyethylene(10)-N-tallow-1 ,3-diaminopropane, Thonzonium bromide, Trimethyl(tetradecyl)ammonium bromide, 1 ,3,5-Triazine-1 ,3,5(2H,4H,6H)- triethanol, 1 -Decanaminium, N-decyl-N, N-dimethyl-, chloride, Didecyl dimethyl ammonium chloride, 2-(2-(p-(Diisobutyl)cresosxy)ethoxy)ethyl dimethyl benzyl ammonium chloride, 2-(2-(p-(Diisobutyl)phenoxy)ethoxy)ethyl dimethyl benzyl ammonium chloride, Alkyl 1 or 3 benzyl-1 -(2-
  • Alkyldimethyl(ethylbenzyl) ammonium chloride (C-i 2- i 8 ), Di-(C 8- io)-alkyl dimethyl ammonium chlorides, Dialkyl dimethyl ammonium chloride, Dialkyl methyl benzyl ammonium chloride, Didecyl dimethyl ammonium chloride, Diisodecyl dimethyl ammonium chloride, Dioctyl dimethyl ammonium chloride, Dodecyl bis (2- hydroxyethyl) octyl hydrogen ammonium chloride, Dodecyl dimethyl benzyl ammonium chloride, Dodecylcarbamoyl methyl dinethyl benzyl ammonium chloride, Heptadecyl hydroxyethylimidazolinium chloride, Hexahydro-1 ,3,5 - tris(2-hydroxyethyl)-s-triazine, Hexahydro-1 ,
  • Exemplary cationic halogen-containing compounds include, but are not limited to, cetylpyridinium halides, cetyltrimethylammonium halides,
  • suitable cationic halogen containing compounds comprise, but are not limited to, cetylpyridinium chloride (CPC), cetyltrimethylammonium chloride,
  • cetylbenzyldimethylammonium chloride cetylpyridinium bromide (CPB), cetyltrimethylammonium bromide (CTAB), cetyidimethylethylammonium bromide, cetyltributylphosphonium bromide, dodecyltrimethylammonium bromide, and tetrad ecyltrimethylammonium bromide.
  • the cationic halogen containing compound is CPC, although the compositions of the present invention are not limited to formulation with an particular cationic containing compound.
  • Suitable anionic surfactants include, but are not limited to, a carboxylate, a sulphate, a sulphonate, a phosphate, chenodeoxycholic acid, chenodeoxycholic acid sodium salt, cholic acid, ox or sheep bile, Dehydrocholic acid, Deoxycholic acid, Deoxycholic acid, Deoxycholic acid methyl ester, Digitonin, Digitoxigenin, ⁇ , ⁇ -Dimethyldodecylamine N-oxide, Docusate sodium salt,
  • Glycochenodeoxycholic acid sodium salt Glycocholic acid hydrate, synthetic, Glycocholic acid sodium salt hydrate, synthetic, Glycodeoxycholic acid
  • Taurolithocholic acid 3-sulfate disodium salt Tauroursodeoxycholic acid sodium salt
  • Trizma ® dodecyl sulfate Trizma ® dodecyl sulfate
  • TWEEN ® 80 Ursodeoxycholic acid, semi-synthetic derivatives thereof, and combinations thereof.
  • Suitable zwitterionic surfactants include, but are not limited to, an N-alkyl betaine, lauryl amindo propyl dimethyl betaine, an alkyl dimethyl glycinate, an N- alkyl amino propionate, CHAPS, minimum 98% (TLC), CHAPS, SigmaUltra, minimum 98% (TLC), CHAPS, for electrophoresis, minimum 98% (TLC),
  • CHAPSO minimum 98%, CHAPSO, SigmaUltra, CHAPSO, for electrophoresis, 3-(Decyldimethylammonio)propanesulfonate inner salt, 3- Dodecyldimethylammonio)propanesulfonate inner salt, SigmaUltra, 3- (Dodecyldimethylammonio)propanesulfonate inner salt, 3-(N,N- Dimethylmyristylammonio)propanesulfonate, 3-(N,N- Dimethyloctadecylammonio)propanesulfonate, 3-(N,N- Dimethyloctylammonio)propanesulfonate inner salt, 3-(N,N- Dimethylpalmitylammonio)propanesulfonate, semi-synthetic derivatives thereof, and combinations thereof.
  • the nanoemulsion RSV vaccine comprises a cationic surfactant, which can be cetylpyridinium chloride. In other embodiments of the invention, the nanoemulsion RSV vaccine comprises a cationic surfactant, and the concentration of the cationic surfactant is less than about 5.0% and greater than about 0.001 %. In yet another embodiment of the invention, the nanoemulsion RSV vaccine comprises a cationic surfactant, and the
  • concentration of the cationic surfactant is selected from the group consisting of less than about 5%, less than about 4.5%, less than about 4.0%, less than about 3.5%, less than about 3.0%, less than about 2.5%, less than about 2.0%, less than about 1 .5%, less than about 1 .0%, less than about 0.90%, less than about 0.80%, less than about 0.70%, less than about 0.60%, less than about 0.50%, less than about 0.40%, less than about 0.30%, less than about 0.20%, or less than about 0.10%.
  • the concentration of the cationic agent in the nanoemulsion vaccine is greater than about 0.002%, greater than about 0.003%, greater than about 0.004%, greater than about 0.005%, greater than about 0.006%, greater than about 0.007%, greater than about 0.008%, greater than about 0.009%, greater than about 0.010%, or greater than about 0.001 %. In one embodiment, the concentration of the cationic agent in the nanoemulsion vaccine is less than about 5.0% and greater than about 0.001 %.
  • the nanoemulsion vaccine comprises at least one cationic surfactant and at least one non-cationic surfactant.
  • the non-cationic surfactant is a nonionic surfactant, such as a polysorbate (Tween), such as polysorbate 80 or polysorbate 20.
  • the non-ionic surfactant is present in a concentration of about 0.01 % to about 5.0%, or the non-ionic surfactant is present in a concentration of about 0.1 % to about 3%.
  • the nanoemulsion vaccine comprises a cationic surfactant present in a concentration of about 0.01 % to about 2%, in combination with a nonionic surfactant.
  • Additional compounds suitable for use in the nanoemulsion RSV vaccines of the invention include but are not limited to one or more solvents, such as an organic phosphate-based solvent, bulking agents, coloring agents,
  • additional compounds can be admixed into a previously emulsified nanoemulsion vaccine, or the additional compounds can be added to the original mixture to be emulsified. In certain of these embodiments, one or more additional compounds are admixed into an existing nanoemulsion composition immediately prior to its use.
  • Suitable preservatives in the nanoemulsion RSV vaccines of the invention include, but are not limited to, cetylpyridinium chloride, benzalkonium chloride, benzyl alcohol, chlorhexidine, imidazolidinyl urea, phenol, potassium sorbate, benzoic acid, bronopol, chlorocresol, paraben esters, phenoxyethanol, sorbic acid, alpha-tocophernol, ascorbic acid, ascorbyl palmitate, butylated
  • metabisulphite citric acid, edetic acid, semi-synthetic derivatives thereof, and combinations thereof.
  • suitable preservatives include, but are not limited to, benzyl alcohol, chlorhexidine (bis (p-chlorophenyldiguanido) hexane),
  • chlorphenesin (3-(-4-chloropheoxy)-propane-1 ,2-diol)
  • Kathon CG methyl and methylchloroisothiazolinone
  • parabens methyl, ethyl, propyl, butyl
  • the nanoemulsion RSV vaccine may further comprise at least one pH adjuster.
  • Suitable pH adjusters in the nanoemulsion vaccine of the invention include, but are not limited to, diethyanolamine, lactic acid, monoethanolamine, triethylanolamine, sodium hydroxide, sodium phosphate, semi-synthetic derivatives thereof, and combinations thereof.
  • the nanoemulsion RSV vaccine can comprise a chelating agent.
  • the chelating agent is present in an amount of about 0.0005% to about 1 %.
  • chelating agents include, but are not limited to, ethylenediamine, ethylenediaminetetraacetic acid (EDTA), phytic acid, polyphosphoric acid, citric acid, gluconic acid, acetic acid, lactic acid, and dimercaprol, and a preferred chelating agent is ethylenediaminetetraacetic acid.
  • the nanoemulsion RSV vaccine can comprise a buffering agent, such as a pharmaceutically acceptable buffering agent.
  • buffering agents include, but are not limited to, 2-Amino-2-methyl-1 ,3-propanediol, >99.5% (NT), 2-Amino-2-methyl-1 -propanol, >99.0% (GC), L-(+)-Tartaric acid, >99.5% (T), ACES, >99.5% (T), ADA, >99.0% (T), Acetic acid, >99.5% (GC/T), Acetic acid, for luminescence, >99.5% (GC/T), Ammonium acetate solution, for molecular biology, ⁇ 5 M in H 2 O, Ammonium acetate, for luminescence, >99.0% (calc.
  • Ammonium sulfate solution for molecular biology, 3.2 M in H 2 O, Ammonium tartrate dibasic solution , 2 M in H 2 O (colorless solution at 20 °C), Ammonium tartrate dibasic, >99.5% (T), BES buffered saline, for molecular biology, 2x concentrate, BES , >99.5% (T), BES, for molecular biology, >99.5% (T), BICINE buffer Solution, for molecular biology, 1 M in H 2 O, BICINE, >99.5% (T), BIS-TRIS, >99.0% (NT), Bicarbonate buffer solution , >0.1 M Na 2 CO 3 , >0.2 M NaHCO 3 , Boric acid , >99.5% (T), Boric acid, for molecular biology, >99.5% (T), CAPS, >99.0% (TLC), CHES, >99.5% (T), Calcium acetate hydrate, >99.0% (calc. on dried material, KT), Calcium
  • T Sodium citrate monobasic, anhydrous, >99.5% (T), Sodium citrate tribasic dihydrate, >99.0% (NT), Sodium citrate tribasic dihydrate, for luminescence, >99.0% (NT), Sodium citrate tribasic dihydrate, for molecular biology, >99.5% (NT), Sodium formate solution, 8 M in H 2 O, Sodium oxalate, >99.5% (RT), Sodium phosphate dibasic dihydrate, >99.0% (T), Sodium phosphate dibasic dihydrate, for luminescence, >99.0% (T), Sodium phosphate dibasic dihydrate , for molecular biology, >99.0% (T), Sodium phosphate dibasic dodecahydrate, >99.0% (T), Sodium phosphate dibasic solution, 0.5 M in H 2 O, Sodium phosphate dibasic, anhydrous, >99.5% (T), Sodium phosphate dibasic , for molecular biology,
  • T TM buffer solution, for molecular biology, pH 7.4, TNT buffer solution, for molecular biology, pH 8.0, TRIS Glycine buffer solution, 10* concentrate, TRIS acetate - EDTA buffer solution, for molecular biology, TRIS buffered saline, 10* concentrate, TRIS glycine SDS buffer solution, for electrophoresis, 10* concentrate, TRIS
  • phosphate-EDTA buffer solution for molecular biology, concentrate, 10* concentrate, T cine, >99.5% (NT), Triethanolamine, >99.5% (GC), Thethylamine, >99.5% (GC), Triethylammonium acetate buffer, volatile buffer, ⁇ 1 .0 M in H 2 O, Triethylammonium phosphate solution, volatile buffer, -1 .0 M in H 2 O,
  • Trimethylammonium acetate solution volatile buffer, ⁇ 1 .0 M in H 2 O,
  • Trimethylammonium phosphate solution volatile buffer, ⁇ 1 M in H 2 O, Tris-EDTA buffer solution, for molecular biology, concentrate, 100* concentrate, Tris-EDTA buffer solution , for molecular biology, pH 7.4, Tris-EDTA buffer solution, for molecular biology, pH 8.0, Trizma ® acetate, >99.0% (NT), Trizma ® base , >99.8% (T), Trizma ® base, >99.8% (T), Trizma ® base , for luminescence, >99.8% (T), Trizma ® base, for molecular biology, >99.8% (T), Trizma ® carbonate, >98.5% (T), Trizma ® hydrochloride buffer solution, for molecular biology, pH 7.2, Trizma ® hydrochloride buffer solution, for molecular biology, pH 7.4, Trizma hydrochloride buffer solution, for molecular biology, pH 7.6, Trizma ®
  • hydrochloride buffer solution for molecular biology, pH 8.0, Trizma ®
  • Trizma ® hydrochloride >99.0% (AT)
  • Trizma ® hydrochloride for luminescence, >99.0% (AT)
  • Trizma ® hydrochloride for molecular biology, >99.0% (AT)
  • Trizma ® maleate >99.5% (NT).
  • the nanoemulsion RSV vaccine can comprise one or more emulsifying agents to aid in the formation of emulsions.
  • Emulsifying agents include compounds that aggregate at the oil/water interface to form a kind of continuous membrane that prevents direct contact between two adjacent droplets.
  • Certain embodiments of the present invention feature nanoemulsion vaccines that may readily be diluted with water or another aqueous phase to a desired concentration without impairing their desired properties.
  • the RSV vaccine can further comprise one or more immune modulators.
  • immune modulators include, but are not limited to, chitosan, glucan, enterotoxin, nucleic acid (CpG motifs), MF59, alum, ASO system, etc. It is within the purview of one of ordinary skill in the art to employ suitable immune modulators in the context of the present invention.
  • An immune modulator can be present in the vaccine composition at any pharmaceutically acceptable amount including, but not limited to, from about 0.001 % up to about 10%, and any amount inbetween, such as about 0.01 %, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1 %, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1 %, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%.
  • compositions that comprise the nanoemulsion RSV vaccine in a therapeutically effective amount and suitable, pharmaceutically-acceptable excipients for pharmaceutically acceptable delivery.
  • excipients are well known in the art.
  • terapéuticaally effective amount it is meant any amount of the nanoemulsion RSV vaccine that is effective in preventing, treating or ameliorating a disease caused by the RSV pathogen associated with the immunogen administered in the composition comprising the nanoemulsion RSV vaccine.
  • protective immune response it is meant that the immune response is associated with prevention, treating, or amelioration of a disease. Complete prevention is not required, though is encompassed by the present invention.
  • the immune response can be evaluated using the methods discussed herein or by any method known by a person of skill in the art.
  • Intranasal administration includes administration via the nose, either with or without concomitant inhalation during administration. Such administration is typically through contact by the composition comprising the nanoemulsion RSV vaccine with the nasal mucosa, nasal turbinates or sinus cavity.
  • Administration by inhalation comprises intranasal administration, or may include oral inhalation. Such administration may also include contact with the oral mucosa, bronchial mucosa, and other epithelia.
  • Exemplary dosage forms for pharmaceutical administration are described herein. Examples include but are not limited to liquids, ointments, creams, emulsions, lotions, gels, bioadhesive gels, sprays, aerosols, pastes, foams, sunscreens, capsules, microcapsules, suspensions, pessary, powder, semi-solid dosage form, etc.
  • the pharmaceutical nanoemulsion RSV vaccines may be formulated for immediate release, sustained release, controlled release, delayed release, or any combinations thereof, into the epidermis or dermis.
  • the formulations may comprise a penetration-enhancing agent.
  • Suitable penetration- enhancing agents include, but are not limited to, alcohols such as ethanol, triglycerides and aloe compositions.
  • the amount of the penetration-enhancing agent may comprise from about 0.5% to about 40% by weight of the formulation.
  • nanoemulsion RSV vaccines of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis. Further, the nanoemulsion RSV vaccines of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis. Further, the nanoemulsion RSV vaccines of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis. Further, the nanoemulsion RSV vaccines of the invention can be applied and/or delivered utilizing electrophoretic delivery/electrophoresis. Further, the
  • composition may be a transdermal delivery system such as a patch or administered by a pressurized or pneumatic device (i.e., "gene gun”).
  • a pressurized or pneumatic device i.e., "gene gun”
  • Such methods which comprise applying an electrical current, are well known in the art.
  • the pharmaceutical nanoemulsion RSV vaccines for administration may be applied in a single administration or in multiple administrations.
  • the nanoemulsion RSV vaccines may be occluded or semi-occluded. Occlusion or semi-occlusion may be performed by overlaying a bandage, polyoleofin film, article of clothing, impermeable barrier, or semi- impermeable barrier to the topical preparation.
  • W 8 o5EC An exemplary nanoemulsion adjuvant composition according to the invention is designated "W 8 o5EC" adjuvant.
  • the composition of W 8 o5EC adjuvant is shown in the table below (Table 1 )H.
  • the mean droplet size for the W 8 o5EC adjuvant is ⁇ 400nm. All of the components of the nanoemulsion are included on the FDA inactive ingredient list for Approved Drug Products.
  • the nanoemulsion adjuvants are formed by emulsification of an oil, purified water, nonionic detergent, organic solvent and surfactant, such as a cationic surfactant.
  • An exemplary specific nanoemulsion adjuvant is designated as "60%W 80 5EC".
  • the 60%W 80 5EC-adjuvant is composed of the ingredients shown in Table 2 below: purified water, USP; soybean oil USP; Dehydrated Alcohol, USP [anhydrous ethanol]; Polysorbate 80, NF and cetylpyridinium chloride, USP (CPC). All components of this exemplary nanoemulsion are included on the FDA list of approved inactive ingredients for Approved Drug Products.
  • the nanoemulsions of the invention can be formed using classic emulsion forming techniques. See e.g., U.S. 2004/0043041 .
  • the oil is mixed with the aqueous phase under relatively high shear forces (e.g., using high hydraulic and mechanical forces) to obtain a nanoemulsion comprising oil droplets having an average diameter of less than about 1000 nm.
  • relatively high shear forces e.g., using high hydraulic and mechanical forces
  • Some embodiments of the invention employ a nanoemulsion having an oil phase comprising an alcohol such as ethanol.
  • the oil and aqueous phases can be blended using any apparatus capable of producing shear forces sufficient to form an emulsion, such as French Presses or high shear mixers (e.g., FDA approved high shear mixers are available, for example, from Admix, Inc., Manchester, N.H.). Methods of producing such emulsions are described in U.S. Pat. Nos. 5,103,497 and 4,895,452, herein incorporated by reference in their entireties.
  • the nanoemulsions used in the methods of the invention comprise droplets of an oily discontinuous phase dispersed in an aqueous continuous phase, such as water or PBS.
  • the nanoemulsions of the invention are stable, and do not deteriorate even after long storage periods.
  • Certain nanoemulsions of the invention are non-toxic and safe when swallowed, inhaled, or contacted to the skin of a subject.
  • compositions of the invention can be produced in large quantities and are stable for many months at a broad range of temperatures.
  • the nanoemulsion can have textures ranging from that of a semi-solid cream to that of a thin lotion, to that of a liquid and can be applied topically by any pharmaceutically acceptable method as stated above, e.g., by hand, or nasal drops/spray.
  • the emulsion may be in the form of lipid structures including, but not limited to, unilamellar, multilamellar, and paucliamellar lipid vesicles, micelles, and lamellar phases.
  • the present invention contemplates that many variations of the described nanoemulsions will be useful in the methods of the present invention.
  • To determine if a candidate nanoemulsion is suitable for use with the present invention three criteria are analyzed. Using the methods and standards described herein, candidate emulsions can be easily tested to determine if they are suitable. First, the desired ingredients are prepared using the methods described herein, to determine if a nanoemulsion can be formed. If a
  • the candidate nanoemulsion should form a stable emulsion.
  • a nanoemulsion is stable if it remains in emulsion form for a sufficient period to allow its intended use. For example, for nanoemulsions that are to be stored, shipped, etc., it may be desired that the nanoemulsion remain in emulsion form for months to years. Typical nanoemulsions that are relatively unstable, will lose their form within a day.
  • the candidate nanoemulsion should have efficacy for its intended use. For example, the emulsions of the invention should kill or disable RSV virus to a detectable level, or induce a protective immune response to a detectable level.
  • the nanoemulsion of the invention can be provided in many different types of containers and delivery systems.
  • the nanoemulsions are provided in a cream or other solid or semi-solid form.
  • the nanoemulsions of the invention may be incorporated into hydrogel formulations.
  • the nanoemulsions can be delivered (e.g., to a subject or customers) in any suitable container. Suitable containers can be used that provide one or more single use or multi-use dosages of the nanoemulsion for the desired application.
  • the nanoemulsions are provided in a suspension or liquid form.
  • Such nanoemulsions can be delivered in any suitable container including spray bottles and any suitable pressurized spray device. Such spray bottles may be suitable for delivering the nanoemulsions intranasally or via inhalation.
  • These nanoemulsion-containing containers can further be packaged with instructions for use to form kits.
  • RSV A2 strain as compared to RSV L19 strain, and Cell Lysate vs. Supernatant.
  • This cell lysate was assayed for quantity of F protein associated with the cells.
  • L19 and A2 virus was extracted and purified from HEP-2 infected cells 4 days following infection. Purified virus was compared for protein contents.
  • Results Normalized samples were assayed in Western blots using a polyclonal anti RSV antibodies. F and G protein contents were compared between L19 and A2 strains. The density of the bands was compared using image capturing and a Kodak software. A mock non-infected cell culture was prepared as a control.
  • Figure 1 shows an SDS PAGE of HRSV Infected Cell Lysate (SDS treated) with L19 and A2
  • Figure 2 shows an SDS-PAGE of L19 and A2 HRSV Cell Lysate (cells & supernatant)
  • Figure 3 shows an SDS PAGE of HRSV L19 and A2 Purified Virus.
  • Table 3 shows comparable HRSV F and G protein from L19 and A2 levels from SDS-PAGE.
  • Table 4 shows comparable HRSV L19 and A2 F and G protein from infected cells (Lysate, Supernatant).
  • Table 5 shows comparable HRSV L19 and A2 F and G protein from SDS PAGE.
  • RSV L19 virus infected cells produce between 3-1 1 fold higher quantities of RSV viral proteins as compared to A2 infected cells.
  • the purpose of this example was to compare F protein expression in Hep- 2 cells infected with different strains of RSV virus (L19 vs. A2) for various infection times (24 hours vs. 4 days).
  • Materials and Methods Hep-2 cells were infected with either L19 or A2 RSV virus. 2 sets of 4 flasks total.
  • C + M Cell + Culture Medium (culture medium reserved).
  • Results The results are detailed in Figure 4, which shows a Western blot of HRSV L19 and A2 F and G protein expression 24 hours after virus infection.
  • Table 8 shows a density analysis of HRSV F and G protein band from Western Blot. Table 8.
  • both cell-associated viral particles and culture media- associated viral particles express much higher G in L19 infected cells compared to those infected with RSV A2 strain.
  • the purpose of this example was to demonstrate the associated of a nanoemulsion with viral antigen.
  • the sections on the grids were stained with saturated uranyl acetate in distilled and deionizer water (pH 7) for 10 minutes followed by lead citrate for 5 minutes.
  • the samples were viewed with a Philips CM-100 TEM equipped with a computer controlled cognitive stage, a high resolution (2K x 2K) digital camera and digitally imaged and captured using X- Stream imaging software (SEM Tech Solutions, Inc., North Billerica, MA).
  • the materials/particles inside the oil core of the droplets that represent the Fluzone ® antigens. Since the antigen is incorporated in the core, and is surrounded by the core material, it is protected from staining by the electron dense stain. This leads to a white counter staining effect in the core. The localization of the antigen within the core shields the antigen-sensitive protein subunits in the emulsion, and may protect the antigen from degradation, and thus enhancing stability. There are very few Fluzone ® particles outside of the NE particles that were stained dark in color (Figs. 6a and 6b).
  • the purpose of this example was to compare several different approaches for inactivation of RSV, including ⁇ -propiolactone and W 8 o5EC Nanoemulsion, via nasal vaccination in a mouse.
  • W 8 o5EC an oil-in-water nanoemulsion with both antiviral and adjuvant activity
  • ⁇ -PL ⁇ -propiolactone
  • the two vaccines were administered intranasally (IN) to BALB/C mice at weeks 0 and 4. Mice were bled prior to dosing and at 3 weeks post-boost and then tested for specific antibodies against F-protein.
  • Animals were challenged nasally with 1 x10 5 pfu RSV L19 at week 8 and checked for airway hyper-reactivity (AHR), lung cytokines, and viral protein mRNA clearance using PCR.
  • AHR airway hyper-reactivity
  • lung cytokines cytokines
  • viral protein mRNA clearance using PCR.
  • ⁇ -PL inactivated RSV virus vaccine is associated with AHR following viral challenge in a mouse model of RSV infection.
  • nanoemulsion viral inactivation produced no AHR and induced a significantly increased IL-17 production and improved viral clearance. This suggests a novel pathway of immune protection that may provide benefit for vaccination against RSV.
  • the purpose of this example is to describe exemplary nanoemulsions useful as adjuvants for an RSV vaccine.
  • a total of 10 nanoemulsion formulations were evaluated in mice and cotton rats.
  • W 80 5EC alone, six W 80 5EC + Poloxamer 407 and Poloxamer 188 (P407 and P188) formulations as well as two W 8 o5EC + Chitosan and one W 8 o5EC + Glucan formulation have been produced and assessed for stability over 2 weeks under accelerated conditions at 40°C (Table 1 ). All 10 nanoemulsions were stable for at least 2 weeks at 40°C.
  • NE188 comprising: (a) CPC/Tween 80/P188 (ratio of 1 :1 :5), (b) Particle size ⁇ 300nm, (c) enhanced mucoadhesiveness (IN), and (d) enhanced residence time (IM) (Table 1 1 ).
  • the purpose of this example is to describe RSV viral strains useful in the vaccines of the invention.
  • NanoBio obtained and evaluated a novel L19 RSV strain to test as an antigen in the nanoemulsion inactivated/nanoemulsion adjuvanted RSV vaccine.
  • This strain was found to cause infection and enhanced respiratory disease (ERD) in mice.
  • ERD enhanced respiratory disease
  • data published showed that it conferred protection without induction of ERD in mice when formulated with nanoemulsion.
  • This L19 strain was compared to a Wildtype A2 strain obtained from the American Type Culture Collection (ATCC).
  • the RSV Strain L19 isolate was isolated from an RSV-infected infant with respiratory illness in Ann Arbor, Michigan on 3 January 1967 in WI-38 cells and passaged in SPAFAS primary chick kidney cells followed by passage in SPAFAS primary chick lung cells prior to transfer to MRC-5 cells (Herlocher 1999) and subsequently Hep2 cells (Lukacs 2006).
  • Comparison of RSV L19 genome (15,191 -nt; GenBank accession number FJ614813) with the RSV strain A2 (15,222-nt; GenBank accession number M74568) shows that 98% of the genomes are identical. Most coding differences between L19 and A2 are in the F and G genes. Amino acid alignment of the two strains showed that F protein has 14 (97% identical) and G protein has 20 (93% identical) amino acid differences.
  • RSV L19 strain has been demonstrated in animal models to mimic human infection by stimulating mucus production and significant induction of IL-13 using an inoculum of 1 x 10 5 plaque forming units (PFU)/mouse by intra-tracheal administration (Lukacs 2006).
  • PFU plaque forming units
  • RSV L19 Strain NanoBio developed and optimized RSV propagation and purification methods for three viral strains grown in Vera cells and has established multiplicity of Infection (MOI), optimized purification and concentration of the antigen using PEG6000 precipitation and ultracentrifugation.
  • MOI multiplicity of Infection
  • the RSV L19 viral strain is unique in that it produces significantly higher yields of F protein (approximately 10-30 fold more per PFU) than the other strains.
  • F protein content may be a key factor in immunogenicity and the L19 strain currently elicits the most robust immune response.
  • the L19 strain has a shorter propagation time and therefore will be more efficient from a manufacturing perspective.
  • NanoBio proposes to produce RSV L19 strain virus for the vaccine in a qualified Vera cell line following single plaque isolation of the L19 strain and purification of the virus to establish a Master Viral Seed Bank and Working Viral Seed Bank. The results comparing the three viral strains are provided in Table 12.
  • the purpose of this example is to describe Inactivation of RSV L19 viral strain with different nanoemulsion adjuvants.
  • the nanoemulsions (1 ) W 8 o5EC, (2) W 80 5EC with P407; (3) W 80 5EC with P188, (4) W 8 o5EC with high and low molecular weight Chitosan, and (5) W 80 5EC with Glucan, have been tested with the RSV L19 viral strain to determine viral inactivation. Inactivation with 20% nanoemulsion was performed for 2 hours at room temperature and with 0.25% ⁇ _ for 16 hours at 4°C followed by 2 hours at 37°C. The treated virus was passaged three times in Hep-2 cells and Western blot analysis was performed on cell lysate to determine presence of live virus. See Figure 7.
  • Figure 7 shows the viral inactivation by Western blot assessment, with lanes containing: (1 ) W 80 5EC (Lane 1 ), (2) W 80 5EC + 0.03% B 1 ,3 Glucan (lane 2), (3) W 8 o5EC + 0.3% Chitosan (medium molecular weight) + acetic acid (lane 3), (4) W 80 5EC + 0.3% P407 (lane 4), (5) W 80 5EC + 0.3%
  • FIG. 7 shows that three consecutive passages of the NE-treated virus in a cell culture resulted in no detected viral antigen when blotted against RSV antibodies in a western blot. This three cell culture passage test is well established and accepted method for determining viral inactivation. Of note, all lanes in Figure 7 have a thick background band, which is not a viral band, but is bovine serum albumin. Viral proteins can be detected only in the positive control (lane 8).
  • Example 8 shows that
  • the purpose of this example was to evaluate the short term stability of RSV vaccines.
  • Target doses of RSV L19 viral preparations were formulated to achieve a final nanoemulsion concentration of 20%.
  • Vaccine was stored at RT and at 4°C. Stability test parameters included physical and chemical analysis (Table 13).
  • the W 80 5EC/P188 (1 :1 :5) and W 80 5EC/P188 (1 :5:1 ) formulations were also tested with a live virus RSV A2 strain as opposed to RSV L19 strain for a maximum of 14 days; the 1 :1 :5 formulation demonstrated stability whereas the 1 :5:1 formulation demonstrated potential agglomeration (Table 14).
  • Figure 8 shows an example of G band intensity of RSV strain L19 with W 8 o5EC +/- ⁇ inactivation by Western blot at day 0 ( Figure 8A) and following 14 days of storage at RT or 4°C ( Figure 8B).
  • Figure 8 shows a Western blot analysis performed with anti-RSV antibody (anti-G); L19 virus 4 x 10 6 PFU/lane, 2 x 10 6 PFU/lane, and 1 x 10 6 PFU/lane +/- ⁇ _ inactivation combined with W 8 o5EC as indicated.
  • Specimens were analyzed fresh (Figure 8A) or after 14 days at 4°C or room temperature (RT) ( Figure 8B).
  • the purpose of this example was to evaluate the immunogenicity of an
  • mice were immunized intramuscularly as shown in Table 15. Mice received 50 ⁇ of RSV adjuvanted vaccine IM at 0 weeks. Mice were bled on 0 and 3 weeks and tested for serum antibodies. Chitosan was used as an immune- modulator to enhance the immune response in addition to the adjuvant activity contributed to the nanoemulsion.
  • FIG. 9 shows the immune response (IgG, pg/ml) at week 3 following vaccination in mice vaccinated IM with different nanoemulsion formulations with and without chitosan: (1 ) RSV strain L19 + 2.5% W 80 5EC + 0.1 % Low Mol. Wt.
  • the purpose of this example was to determine the immunogenicity of RSV vaccines according to the invention in Cotton rats.
  • Cotton rats are the accepted animal species for evaluating immunogenicity and efficacy of RSV vaccines. Using data generated in mice, two nanoemulsions were selected for evaluation in Cotton rats. The two initial formulations studied include the W 80 5EC and the W 80 Pi 88 5EC (1 :1 :5) (see Tables 10 and 1 1 above).
  • ELISA Unit/ uq/ml The amount of specific antibody to F protein was calculated by area under the curve in the ELISA in relation to a defined reference serum which was assigned an arbitrary 100 EU.
  • Example 11 The amount of specific antibody to F protein was calculated by area under the curve in the ELISA in relation to a defined reference serum which was assigned an arbitrary 100 EU.
  • the purpose of this example was to determine the effect of RSV vaccines according to the invention on neutralizing antibodies, as well as cross-reactivity of an RSV vaccine comprising RSV strain L19 against other RSV strains following IN administration.
  • NEU Neutralization units
  • Figure 13 shows neutralizing antibody titers at 6 weeks time point (Figure 13A). It is noteworthy that all animals vaccinated with 3.2x10 6 PFU RSV strain L19 inactivated with 60% W 80 5EC or 60% W 80 Pi 88 5EC generated robust neutralizing antibodies. There is a statistically significant positive
  • NU Neutralization Unit
  • NU/EU Viral neutralizing antibody antibodies (NU) per the one EU F-protein antibody ( Figure 13B)
  • Figure 14 shows neutralizing antibodies on day 4 and day 8.
  • Figure 14A shows the results for W 80 Pi 88 5EC nanoemulsion combined with RSV strain L19
  • Figure 14B shows the results for W 80 5EC nanoemulsion combined with RSV strain L19.
  • All cotton rats demonstrated high neutralizing antibodies (NU) against the vaccine RSV strain L19. Neutralizing antibodies were rising steadily following the challenge (Y axis).
  • Day 8 neutralizing units (NU) were higher than Day 4 NU. Na ' fve Cotton Rats did not show any neutralization activity in their sera. Serum neutralizing antibodies and specific activity showed a trend to increase from Day 4 to Day 8 post-challenge.
  • Figure 15 shows the Specific activity of serum antibodies showed that the specific activity (Neutralizing units/ELISA units) of the serum antibodies tends to increase on Day 8 when compared to Day 4 post-challenge.
  • Figure 15A shows the results for W 8 oP-i 88 5EC nanoemulsion combined with RSV strain L19 (NU/EU for the Y axis), at Day 4 and Day 8.
  • Figure 15B shows the results for W 8 o5EC nanoemulsion combined with RSV strain L19 (NU/EU for the Y axis), at Day 4 and Day 8. All cotton rats demonstrated high neutralization activity (Figure 15).
  • Serum of vaccinated cotton rats showed cross protection against RSV strain A2 (in addition to RSV strain L19) on Day 4 post-challenge (Figure 16). Specifically, Figure 16 shows cross protection at Day 4 for cotton rats that received 3 doses of RSV L19 adjuvanted vaccine, then challenged with RSV strain A2.
  • Figure 16A shows the results for W 8 oP-i 88 5EC nanoemulsion combined with RSV strain L19
  • Figure 16B shows the results for W 8 o5EC nanoemulsion combined with RSV strain L19.
  • Serum neutralization activity shows equivalent NU against RSV strain L19 or RSV strain A2, demonstrating cross protection between the two RSV strains.
  • Figure 17 shows viral clearance at Day 4 in the lungs of the cotton rats, by measurement of the RSV strain A2 viral titer (PFU/g) in the lungs of the tested cotton rats.
  • Vaccinated cotton rats (vaccinated with W 80 Pi 88 5EC nanoemulsion combined with RSV strain L19, and W 80 5EC nanoemulsion combined with RSV strain L19), showed complete clearance of RSV strain A2 challenged virus from the lungs of the cotton rats.
  • na ' fve animals shows >10 3 pfu RSV strain A2 /gram of lung (limit of detection was 2.1 x 10 1 pfu/g).
  • Example 12 The purpose of this example was to evaluate Intramuscular vaccination of RSV vaccines according to the invention in Cotton Rats.
  • Cotton rats were vaccinated IM according to the schedule shown in Figure 18.
  • Animals received 50 ⁇ RSV adjuvanted RSV vaccine containing 3.3 g F- protein (20% W 80 5EC nanoemulsion mixed with 1 .6 x 10 5 PFU RSV strain L19 containing 3.3 g F protein).
  • Cotton rats produced a specific immune response against RSV.
  • the antibody levels were diminished until a second boost was administered on week 14. There was a slight increase in the antibody levels following the challenge (Figures 19 and 20).
  • Figure 19 shows the serum immune response in the vaccinated cotton rats.
  • the Y axis shows IgG, g/mL, over a 14 week period, at day 4 post-challenge, and at day 8 post- challenge.
  • Figure 20 shows the serum immune response in the vaccinated cotton rats.
  • Figure 20A shows the end point titers (Y axis) over a 14 week period, at day 4 post-challenge, and at day 8 post-challenge.
  • Figure 20B shows the ELISA units (Y axis) over a 14 week period, at day 4 post-challenge, and at day 8 post-challenge.
  • the efficacy of IM immunization was assessed by challenging the animals with a live A2 strain of RSV, which is a strain that causes disease in humans.
  • a dose of 5x10 5 pfu of RSV strain A2 was administered to animals two weeks after booster immunization of the RSV L19 nanoemulsion-adjuvanted vaccine.
  • a na ' fve age-matched group was also challenged. Half of the animals in each group were sacrificed on day 4 post challenge, at which time the maximum viral load was demonstrated in the lungs of Cotton Rats. The other half were sacrificed at day 8.
  • Viral clearance Lung culture showed that all vaccinated animals had no virus in their lungs at 4 days post challenge while na ' fve animals had virus loads of 10 3 pfu RSV strain A2/g of lung tissue ( Figure 21 ). Specifically, Figure 21 shows viral clearance at Day 4 in the lungs of the cotton rats, by measurement of the RSV strain A2 viral titer (PFU/g) in the lungs of the tested cotton rats.
  • the antibodies generated are highly effective in neutralizing live virus and there is a linear relationship between neutralization and antibody titers. Furthermore, antibodies generated in cotton rats showed cross protection when immunized with the RSV L19 strain and challenged with the RSV A2 strain. Both IM and IN immunization established memory that can be invoked or recalled after an exposure to antigen either as a second boost or exposure to live virus. Example 13.
  • the purpose of this example was to compare intranasal (IN) versus intramuscular (IM) administration of a W 8 o5EC nanoemulsion adjuvanted RSV vaccine.
  • RSV vaccine containing 2x10 5 plaque forming units (PFU) of L19 RSV virus with 1 .7 g of F protein was inactivated with 20% W 8 o5EC nanoemulsion adjuvant.
  • Cells from spleens, cervical and intestinal lymph nodes (LN) were analyzed for RSV-specific cytokines ( Figure 23). Mice were challenged oropharyngeally with 5x10 5 PFU L19 at 8 weeks. Airway hyperreactivity was assessed by plethysmography.
  • Lungs were analyzed day 8 post challenge to assess mRNA of cytokines, viral proteins, and histopathology.
  • mice vaccinated IM had higher anti-F antibodies (GM 396 [95% CI 240-652] vs. 2 [95% CI 0-91 ]) ( Figure 22) and generated more IL-4 and IL-13, after challenge (p ⁇ 0.05) compared to mice vaccinated IN ( Figure 23).
  • IL-17 from spleen cells, cervical LN and intestinal LN was higher after IN vs IM vaccination (GM: 57 vs 1 , 1 19 vs 3 and 51 vs 4 pg/mL, respectively, p ⁇ 0.05) (Figure 23).
  • Figure 24 shows measurement of the cytokines IL-4, IL-13, and IL- 17 in lung tissue following either IN or IM vaccination.
  • IL-4 and IL-13 were expressed at higher amounts following IM administration, with IL-17 showing greater expression following IN administration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Biophysics (AREA)
  • Inorganic Chemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pulmonology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Dermatology (AREA)
  • Biotechnology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)

Abstract

La présente invention concerne des vaccins contre RSV et des procédés pour induire une réponse immunitaire au RSV chez un sujet, comprenant l'administration d'un vaccin RSV.
PCT/US2012/045769 2011-07-06 2012-07-06 Vaccin contre le virus syncytial respiratoire humain WO2013006797A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2014519070A JP2014520805A (ja) 2011-07-06 2012-07-06 ヒト呼吸器合胞体ウイルスワクチン
CA2840982A CA2840982C (fr) 2011-07-06 2012-07-06 Vaccin contre le virus syncytial respiratoire humain
EP12735418.1A EP2729169A1 (fr) 2011-07-06 2012-07-06 Vaccin contre le virus syncytial respiratoire humain

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161504989P 2011-07-06 2011-07-06
US61/504,989 2011-07-06
US201161548248P 2011-10-18 2011-10-18
US61/548,248 2011-10-18

Publications (1)

Publication Number Publication Date
WO2013006797A1 true WO2013006797A1 (fr) 2013-01-10

Family

ID=47437470

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/045769 WO2013006797A1 (fr) 2011-07-06 2012-07-06 Vaccin contre le virus syncytial respiratoire humain

Country Status (5)

Country Link
US (1) US9492525B2 (fr)
EP (1) EP2729169A1 (fr)
JP (3) JP2014520805A (fr)
CA (1) CA2840982C (fr)
WO (1) WO2013006797A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2753354A1 (fr) * 2011-09-09 2014-07-16 Nanobio Corporation Vaccin sous-unitaire contre le virus respiratoire syncytial (rsv) en nano-émulsion
CN104906056A (zh) * 2015-05-15 2015-09-16 南方医科大学 一种用于治疗鼻炎的分散片及其制备方法
US9492525B2 (en) 2011-07-06 2016-11-15 Nanobio Corporation Human respiratory syncytial virus vaccine
US11564392B2 (en) 2017-09-18 2023-01-31 Bayer Healthcare Llc Methods of inactivation of viruses using n-methlyglucamide and its derivatives

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3798310A1 (fr) 2013-03-14 2021-03-31 Emory University Rsv recombiné avec des mutations silencieuses, vaccins et procédés apparentés
US20180273909A1 (en) * 2015-09-22 2018-09-27 Sanofi Pasteur Biologics, Llc Purification of respiratory syncytial virus
EP3368547A4 (fr) 2015-10-29 2019-06-05 Emory University Virus rsv chimère, compositions immunogènes et procédés d'utilisation
CN105875210A (zh) * 2016-06-23 2016-08-24 广西玉林天地网农业科技有限公司 一种佛手柑的嫁接方法
JP7317796B2 (ja) * 2017-07-24 2023-07-31 ノババックス,インコーポレイテッド 呼吸器疾患を治療するための方法および組成物
MX2021005767A (es) 2018-11-15 2021-09-21 Bluewillow Biologics Inc Composiciones de nanoemulsiones que tienen una permeabilidad mejorada.

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4826687A (en) 1985-06-06 1989-05-02 National Institute Of Health Influenza vaccine
US4895452A (en) 1988-03-03 1990-01-23 Micro-Pak, Inc. Method and apparatus for producing lipid vesicles
US5103497A (en) 1989-11-14 1992-04-07 Hicks John W Flying spot endoscope
WO1998043668A1 (fr) * 1997-04-03 1998-10-08 The Regents Of The University Of Michigan Virus syncytial respiratoire attenue
US5962298A (en) 1994-01-11 1999-10-05 Vlaams Interuniversitair Instituut Voor Biotechnologie Influenza vaccine
US6015832A (en) 1997-12-31 2000-01-18 The Regents Of The University Of Michigan Methods of inactivating bacteria including bacterial spores
US6194546B1 (en) 1987-02-10 2001-02-27 Glaxo Wellcome Inc. Fusion proteins
US6506803B1 (en) 1999-04-28 2003-01-14 Regents Of The University Of Michigan Methods of preventing and treating microbial infections
US6559189B2 (en) 1999-04-28 2003-05-06 Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6635676B2 (en) 1999-04-28 2003-10-21 Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US20040043041A1 (en) 1999-04-28 2004-03-04 The Regents Of The University Of Michigan Antimicrobial compositions and methods of use
US6861244B2 (en) 1999-02-11 2005-03-01 Baxter Healthcare S.A. Inactivated influenza virus vaccine for nasal or oral application
US20050208083A1 (en) 2003-06-04 2005-09-22 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositons, and methods of use thereof
US20070036831A1 (en) 2005-08-09 2007-02-15 Nanobio Corporation Nanoemulsion compositions having anti-inflammatory activity
US20070054834A1 (en) 2005-04-11 2007-03-08 Nanobio Corporation Quaternary ammonium halides for treatment of infectious conditions
US7192595B2 (en) 1998-11-30 2007-03-20 Yeda Research And Development Co. Ltd. Peptide-based vaccine for influenza
US7314624B2 (en) 2001-06-05 2008-01-01 The Regents Of The University Of Michigan Nanoemulsion vaccines

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ512980A (en) 1998-12-17 2003-07-25 Aventis Pasteur Multivalent immunogenic composition containing RSV subunit composition and influenza virus preparation
WO2008137747A1 (fr) 2007-05-02 2008-11-13 The Regents Of The University Of Michigan Compositions thérapeutiques à base de nanoémulsion et leurs procédés d'utilisation
EP2349209A2 (fr) * 2008-09-26 2011-08-03 Nanobio Corporation Compositions thérapeutiques de nanoémulsion et procédés d'utilisation de celles-ci
ES2566646T3 (es) * 2009-06-16 2016-04-14 The Regents Of The University Of Michigan Vacunas en nanoemulsión
EP2729169A1 (fr) 2011-07-06 2014-05-14 Nanobio Corporation Vaccin contre le virus syncytial respiratoire humain

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4826687A (en) 1985-06-06 1989-05-02 National Institute Of Health Influenza vaccine
US6194546B1 (en) 1987-02-10 2001-02-27 Glaxo Wellcome Inc. Fusion proteins
US4895452A (en) 1988-03-03 1990-01-23 Micro-Pak, Inc. Method and apparatus for producing lipid vesicles
US5103497A (en) 1989-11-14 1992-04-07 Hicks John W Flying spot endoscope
US5962298A (en) 1994-01-11 1999-10-05 Vlaams Interuniversitair Instituut Voor Biotechnologie Influenza vaccine
WO1998043668A1 (fr) * 1997-04-03 1998-10-08 The Regents Of The University Of Michigan Virus syncytial respiratoire attenue
US6015832A (en) 1997-12-31 2000-01-18 The Regents Of The University Of Michigan Methods of inactivating bacteria including bacterial spores
US7192595B2 (en) 1998-11-30 2007-03-20 Yeda Research And Development Co. Ltd. Peptide-based vaccine for influenza
US6861244B2 (en) 1999-02-11 2005-03-01 Baxter Healthcare S.A. Inactivated influenza virus vaccine for nasal or oral application
US6559189B2 (en) 1999-04-28 2003-05-06 Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US20040043041A1 (en) 1999-04-28 2004-03-04 The Regents Of The University Of Michigan Antimicrobial compositions and methods of use
US6635676B2 (en) 1999-04-28 2003-10-21 Regents Of The University Of Michigan Non-toxic antimicrobial compositions and methods of use
US6506803B1 (en) 1999-04-28 2003-01-14 Regents Of The University Of Michigan Methods of preventing and treating microbial infections
US7314624B2 (en) 2001-06-05 2008-01-01 The Regents Of The University Of Michigan Nanoemulsion vaccines
US20050208083A1 (en) 2003-06-04 2005-09-22 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositons, and methods of use thereof
US20060251684A1 (en) 2003-06-04 2006-11-09 Nanobio Corporation Compositions for inactivating pathogenic microorganisms, methods of making the compositions, and methods of use thereof
US20070054834A1 (en) 2005-04-11 2007-03-08 Nanobio Corporation Quaternary ammonium halides for treatment of infectious conditions
US20070036831A1 (en) 2005-08-09 2007-02-15 Nanobio Corporation Nanoemulsion compositions having anti-inflammatory activity

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
BIELINSKA ET AL., AIDS RESEARCH AND HUMAN RETROVIRUSES, vol. 24, no. 2, 2008, pages 271 - 81
BIELINSKA ET AL., CLIN. VACCINE IMMUNOL., vol. 15, no. 2, 2008, pages 348 - 58
BIELINSKA ET AL., INFECT. IMMUN., vol. 75, no. 8, 2007, pages 4020 - 9
BIELINSKA ET AL.: "A Novel, Killed-Virus Nasal Vaccinia Virus Vaccine", CLIN. VACCINE IMMUNOL., vol. 15, no. 2, 2008, pages 348 - 58, XP008138878, DOI: doi:10.1128/CVI.00440-07
BIELINSKA ET AL.: "Mucosal Immunization with a Novel Nanoemulsion-Based Recombinant Anthrax Protective Antigen Vaccine Protects against Bacillus anthracis Spore Challenge", INFECT IMMUN., vol. 75, no. 8, 2007, pages 4020 - 9, XP055114282, DOI: doi:10.1128/IAI.00070-07
BIELINSKA ET AL.: "Nasal Immunization with a Recombinant HIV gp120 and Nanoemulsion Adjuvant Produces Th1 Polarized Responses and Neutralizing Antibodies to Primary HIV Type 1 Isolates", AIDS RESEARCH AND HUMAN RETROVIRUSES, vol. 24, no. 2, 2008, pages 271 - 81, XP008138884, DOI: doi:10.1089/aid.2007.0148
DENNIS M. LINDELL ET AL: "A Novel Inactivated Intranasal Respiratory Syncytial Virus Vaccine Promotes Viral Clearance without Th2 Associated Vaccine-Enhanced Disease", PLOS ONE, vol. 6, no. 7, 15 July 2011 (2011-07-15), pages e21823, XP055038673, DOI: 10.1371/journal.pone.0021823 *
GOMEZ, M.; MUFSON, M. ET AL.: "Phase-I study MEDI-534, of a live attenuated intranasal vaccine against respiratory syncytial virus and parainfluenza-3 virus in seropositive children", PED INFECT J., vol. 28, 2009, pages 655 - 658
GRAHAM, B.: "Biological challenges and technological opportunities for respiratory syncytial virus vaccine development", IMMUNOLOGICAL REVIEWS, vol. 239, 2011, pages 149 - 166, XP055008499, DOI: doi:10.1111/j.1600-065X.2010.00972.x
HACKING, D.; HULL, J.: "Respiratory syncytial virus - virus biology and host response", J. INFECTION, vol. 45, 2002, pages 18 - 24, XP002961522, DOI: doi:10.1053/jinf.2002.1015
HAMOUDA ET AL.: "A Novel Nanoemulsion Adjuvant Enhancing The Immune Response from Intranasal Influenza Vaccine in Mice in National Foundation for Infectious Disease", 11TH ANNUAL CONFERENCE ON VACCINE RESEARCH, 2008
HERLOCHER ET AL.: "immunological properties of plaque purified strains of live attenuated respiratory syncytial virus (RSV) for human vaccine", VACCINE, vol. 17, no. 2, 1999, pages 172 - 81, XP004139973, DOI: doi:10.1016/S0264-410X(98)00155-8
HERLOCHER M L ET AL: "Immunological properties of plaque purified strains of live attenuated respiratory syncytial virus (RSV) for human vaccine", VACCINE, ELSEVIER LTD, GB, vol. 17, no. 2, 1 January 1999 (1999-01-01), pages 172 - 181, XP004139973, ISSN: 0264-410X, DOI: 10.1016/S0264-410X(98)00155-8 *
HUANG K.; LAWLOR, H.: "Recombinant respiratory syncytial virus F protein expression is hindered by inefficient nuclear export and mRNA processing.", VIRUS GENES, vol. 40, 2010, pages 212 - 221, XP019775273, DOI: doi:10.1007/s11262-010-0449-8
KIM, S.; J JANG. ET AL.: "Single mucosal immunization of recombinant adenovirus-based vaccine expressing F1 protein fragment induces protective mucosal immunity against respiratory syncytial virus infection", VACCINE, vol. 28, 2010, pages 3801 - 3808, XP027057876
KRUIJENS D.; SCHIJF, M.: "Local innate and adaptive immune responses regulate inflammatory cell influx into the lungs after vaccination with formalin inactivated RSV", VACCINE, vol. 29, 2011, pages 2730 - 2741, XP028172356, DOI: doi:10.1016/j.vaccine.2011.01.087
LANGLEY, J.; SALES V. ET AL.: "A dose ranging study of a subunit Respiratory Syncytial Virus subtype A vaccine with and without aluminum phosphate adjuvantation in adults ? 65 years of age", VACCINE, vol. 27, 2009, pages 5913 - 5919, XP026583871, DOI: doi:10.1016/j.vaccine.2009.07.038
LUKACS ET AL.: "Differential immune responses and pulmonary pathophysiology are induced by two different strains of respiratory syncytial virus", AM J PATHOL, vol. 169, no. 3, 2006, pages 977 - 86, XP055160826, DOI: doi:10.2353/ajpath.2006.051055
M. L. MOORE ET AL: "A Chimeric A2 Strain of Respiratory Syncytial Virus (RSV) with the Fusion Protein of RSV Strain Line 19 Exhibits Enhanced Viral Load, Mucus, and Airway Dysfunction", JOURNAL OF VIROLOGY, vol. 83, no. 9, 11 February 2009 (2009-02-11), pages 4185 - 4194, XP055038683, ISSN: 0022-538X, DOI: 10.1128/JVI.01853-08 *
MAKIDON ET AL.: "Pre-Clinical Evaluation of a Novel Nanoemulsion-Based Hepatitis B Mucosal Vaccine", PLOS ONE, vol. 3, no. 8, 2008, pages 2954, XP002676516, DOI: doi:10.1371/JOURNAL.PONE.0002954
MCLELLAN, J.; Y YANG ET AL.: "Structure of the Respiratory Syncytial Virus Fusion glycoprotein in the post-fusion conformation reveals preservation of neutralizing epitopes", J VIROLOGY, 2011
MYC ET AL.: "Development of immune response that protects mice from viral pneumonitis after a single intranasal immunization with influenza A virus and nanoemulsion", VACCINE, vol. 21, no. 25-26, 2003, pages 3801 - 14, XP004446155, DOI: doi:10.1016/S0264-410X(03)00381-5
NALLET, S.; AMACKER, M. ET AL., RESPIRATORY SYNCYTIAL VIRUS SUBUNIT VACCINE BASED ON A RECOMBINANT FUSION PROTEIN EXPRESSED TRANSIENTLY IN MAMMALIAN CELLS, vol. 27, 2009, pages 6415 - 6419
See also references of EP2729169A1
SINGH, S.; DENNIS, V.: "Immuogenicity and efficacy of recombinant RSV-F vaccine in a mouse model", VACCINE, vol. 25, 2007, pages 6211 - 6223, XP022168621, DOI: doi:10.1016/j.vaccine.2007.05.068
SWANSON, K.; SETTEMBRE, E.: "Structural basis for immunization with postfusion respiratory syncytial virus fusion F glycoprotein (RSV F) to elicit high neutralizing antibody titers", PNAS, vol. 108, 2011, pages 9619 - 9624, XP055032519, DOI: doi:10.1073/pnas.1106536108
THARWAT F. TADROS: "Applied Surfactants: Principles and Applications", 2005, WILEY-VCH VERLAG GMBH & CO.
WARREN ET AL.: "Pharmacological and Toxicological Studies on Cetylpyridinium Chloride, A New Germicide", J. PHARMACOL. EXP. THER., vol. 74, 1942, pages 401 - 8

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492525B2 (en) 2011-07-06 2016-11-15 Nanobio Corporation Human respiratory syncytial virus vaccine
EP2753354A1 (fr) * 2011-09-09 2014-07-16 Nanobio Corporation Vaccin sous-unitaire contre le virus respiratoire syncytial (rsv) en nano-émulsion
EP2753354A4 (fr) * 2011-09-09 2015-04-15 Nanobio Corp Vaccin sous-unitaire contre le virus respiratoire syncytial (rsv) en nano-émulsion
US9561271B2 (en) 2011-09-09 2017-02-07 Nanobio Corporation Nanoemulsion respiratory syncytial virus (RSV) subunit vaccine
EP3488863A1 (fr) * 2011-09-09 2019-05-29 Nanobio Corporation Vaccin sous-unitaire contre le virus respiratoire syncytial (rsv) en nano-émulsion
US10596251B2 (en) 2011-09-09 2020-03-24 Nanobio Corporation Nanoemulsion respiratory syncytial virus (RSV) subunit vaccine
CN104906056A (zh) * 2015-05-15 2015-09-16 南方医科大学 一种用于治疗鼻炎的分散片及其制备方法
US11564392B2 (en) 2017-09-18 2023-01-31 Bayer Healthcare Llc Methods of inactivation of viruses using n-methlyglucamide and its derivatives

Also Published As

Publication number Publication date
JP6580093B2 (ja) 2019-09-25
US20130011443A1 (en) 2013-01-10
JP2014520805A (ja) 2014-08-25
CA2840982C (fr) 2022-06-07
JP2019142905A (ja) 2019-08-29
JP2018008968A (ja) 2018-01-18
EP2729169A1 (fr) 2014-05-14
US9492525B2 (en) 2016-11-15
CA2840982A1 (fr) 2013-01-10

Similar Documents

Publication Publication Date Title
US9492525B2 (en) Human respiratory syncytial virus vaccine
US10525121B2 (en) Nanoemulsion influenza vaccine
US10596251B2 (en) Nanoemulsion respiratory syncytial virus (RSV) subunit vaccine
US20220105172A1 (en) Herpes simplex virus nanoemulsion vaccines and methods of using the same
US20200246449A1 (en) Intravenous immunoglobulin compositions specific for respiratory syncytial virus and methods of making and using the same
US20120003277A1 (en) Nanoemulsion vaccines
US20200138935A1 (en) Chlamydia nanoemulsion vaccine

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12735418

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2840982

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014519070

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012735418

Country of ref document: EP